PKAR-I alpha Human

Protein Kinase A regulatory subunit-1 alpha Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of PKAR-I Alpha Human

PKAR-I alpha (cAMP-dependent protein kinase type I-alpha regulatory subunit) is a critical regulatory component of Protein Kinase A (PKA), a central enzyme in cAMP-mediated signaling pathways. This protein, encoded by the PRKAR1A gene (NCBI GeneID: 5573), functions as a dimeric 86 kDa complex composed of two 43 kDa monomers (381 amino acids each) . It serves as a tumor suppressor and plays a key role in modulating cellular responses to extracellular signals by regulating PKA activity .

Key Features

  • Molecular Weight: 86 kDa (dimer), 43 kDa (monomer) .

  • Source: Recombinant protein expressed in Escherichia coli .

  • Purity: >95% (verified by SDS-PAGE) .

  • Formulation: 0.8 mg/mL in 20 mM MOPS (pH 7.0), 150 mM NaCl, 1 mM 2-mercaptoethanol, and 50% glycerol .

  • Stability: Stable at 4°C for 2–4 weeks; long-term storage at -20°C .

Functional Mechanism

PKAR-I alpha inhibits the catalytic subunit (C) of PKA by binding to it in the absence of cAMP. Upon cAMP elevation, conformational changes in PKAR-I alpha—particularly at Asp170 within the cAMP-binding domain (CBD-A)—trigger the release of the catalytic subunit, enabling PKA to phosphorylate downstream targets . The D170A mutation reduces negative cooperativity between cAMP and C-subunit binding, destabilizing the R:C complex .

Genetic Profile

PropertyDetails
Gene NamePRKAR1A (NCBI Official Symbol)
Chromosomal Location17q24.2
UniProt IDP10644
Protein FamilycAMP-dependent protein kinase regulatory subunits
Key DomainscAMP-binding domains (CBD-A and CBD-B), dimerization/docking domain .

Disease Links

Mutations in PRKAR1A are implicated in:

  • Carney Complex Type 1 (CNC1): A multiple neoplasia syndrome featuring cardiac myxomas, endocrine tumors, and skin pigmentation .

  • Acrodysostosis 1: A skeletal dysplasia with hormone resistance .

  • Primary Pigmented Nodular Adrenocortical Disease (PPNAD) .

Functional Roles in Cellular Processes

PKAR-I alpha regulates diverse biological pathways:

  • Signal Transduction: Mediates cAMP-dependent activation of PKA, influencing transcription, metabolism, and cell proliferation .

  • DNA Replication: Interacts with Replication Factor C (RFC2), suggesting a role in nuclear transport during DNA synthesis .

  • Immune Response: Modulates innate immunity via PKA’s interaction with inflammatory pathways .

Experimental Use

  • Inhibitory Activity: Reversibly blocks PKA’s catalytic subunit; inhibition is cAMP-dependent .

  • Structural Studies: Used to investigate cAMP-induced conformational changes and allosteric regulation .

  • Disease Modeling: Employed to study PRKAR1A mutations in Carney complex and related disorders .

Key Research Insights

  • Asp170 Mutagenesis: The D170A mutation disrupts cAMP relay to the R:C interface, reducing PKA activation efficiency .

  • Tissue-Specific Expression: PKAR-I alpha exhibits dynamic expression in tissues like the liver and adrenal glands, correlating with its role in hormone signaling .

Interaction Networks and Pathways

PKAR-I alpha participates in:

  • cAMP-PKA Signaling: Central to metabolic regulation, gene expression, and cell cycle control .

  • Protein-Protein Interactions: Binds RFC2, BRCA1, and GILZ, linking PKA to DNA repair and immune modulation .

Future Directions

Current research focuses on:

  • Therapeutic Targeting: Developing small molecules to modulate PKAR-I alpha in cancers and endocrine disorders .

  • Gene Editing: CRISPR-based studies to repair PRKAR1A mutations in Carney complex .

Product Specs

Introduction
The Protein Kinase A (PKA) Regulatory (R) subunit functions by inhibiting the kinase activity of the Catalytic (C) subunit, effectively shielding it from interacting with physiological substrates. This inhibitory mechanism is reversed when extracellular signals trigger an increase in cytoplasmic cAMP levels. Upon binding to the R subunit, cAMP induces an allosteric change that releases the C subunit, thereby activating a range of downstream signaling cascades. Crystallographic studies have revealed a series of conformational changes within the cyclic nucleotide-binding domain A (CBD-A) of the R subunit that are essential for transmitting the cAMP signal from its binding site to the R:C interaction interface. Notably, Asp170 within CBD-A has been identified as a critical cAMP relay site. The D170A mutation specifically disrupts the negative cooperativity between the cAMP- and C-recognition sites, reducing the KD for the R:C complex in the presence of cAMP by over 12-fold, without significantly affecting the high affinity of R for either binding partner.
Description
Recombinant human PKA regulatory subunit I alpha is a dimeric protein with a molecular weight of 86 kDa (monomer: 381 amino acids, 43 kDa). It undergoes purification using proprietary chromatographic techniques.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
PKA regulatory subunit-I alpha is supplied at a concentration of 0.8 mg/ml in a buffer consisting of 20mM MOPS (pH 7.0), 150mM NaCl, 1mM 2-mercaptoethanol, and 50% glycerol.
Stability
For optimal storage, PKAR-Ia should be kept at 4°C if the entire vial will be used within 2-4 weeks. For long-term storage, storing at -20°C is recommended. Repeated freezing and thawing should be avoided.
Purity
Purity exceeds 95% as determined by SDS-PAGE analysis.
Inhibitory Activity
The cAMP-dependent protein kinase, regulatory subunit RIa, exhibits reversible inhibition of the catalytic subunit Ca of cAMP-dependent protein kinase PKA. This inhibitory effect can be neutralized by the introduction of the second messenger, cAMP.
Synonyms
cAMP-dependent protein kinase type I-alpha regulatory subunit, Tissue-specific extinguisher 1, TSE1, PRKAR1A PKR1, PRKAR1, CAR, CNC, CNC1, PKR1, ADOHR, PPNAD1, ACRDYS1.
Source
Escherichia Coli.
Antigen Amino Acid Sequence

MESGSTAASE EARSLRECEL YVQKHNIQAL LKDSIVQLCT ARPERPMAFL REYFERLEKE
EAKQIQNLQK AGTRTDSRED EISPPPPNPV VKGRRRRGAI SAEVYTEEDA ASYVRKVIPK
DYKTMAALAK AIEKNVLFSH LDDNERSDIF DAMFSVSFIA GETVIQQGDE GDNFYVIDQG
ETDVYVNNEW ATSVGEGGSF GELALIYGTP RAATVKAKTN VKLWGIDRDS YRRILMGSTL
RKRKMYEEFL SKVSILESLD KWERLTVADA LEPVQFEDGQ KIVVQGEPGD EFFIILEGSA
AVLQRRSENE EFVEVGRLGP SDYFGEIALL MNRPRAATVV ARGPLKCVKL DRPRFERVLG
PCSDILKRNI QQYNSFVSLS V

Q&A

What is PKAR-I alpha and what is its role in cellular signaling?

PKAR-I alpha, formally known as cAMP-dependent protein kinase type I-alpha regulatory subunit, is an enzyme encoded by the PRKAR1A gene in humans. It functions as a critical component in the cAMP signaling pathway, where it serves as one of the regulatory subunits of protein kinase A (PKA). The inactive PKA holoenzyme exists as a tetramer composed of two regulatory and two catalytic subunits. When cellular cAMP levels increase, cAMP binds to the regulatory subunits, causing the dissociation of the inactive holoenzyme into a dimer of regulatory subunits (bound to four cAMP molecules) and two free monomeric catalytic subunits . This dissociation activates the catalytic subunits, enabling them to phosphorylate downstream target proteins and transduce cellular signals.

PKAR-I alpha specifically functions as a tissue-specific extinguisher that downregulates the expression of certain liver genes in hepatoma x fibroblast hybrids . The regulatory mechanism involves PKAR-I alpha reversibly inhibiting the catalytic subunit (Cα) of PKA, with this inhibition being reversed through the addition of cAMP as a second messenger .

How do mutations in PRKAR1A contribute to the pathogenesis of Carney complex?

Functional null mutations in the PRKAR1A gene are the primary genetic cause of Carney complex (CNC), an autosomal dominant multiple neoplasia syndrome. The pathogenic mechanism involves dysregulation of PKA signaling. When PRKAR1A is mutated, the normal inhibitory control of the PKA catalytic subunits is compromised, leading to constitutive activation of PKA signaling pathways .

This aberrant PKA activity results in uncontrolled cellular growth and proliferation, particularly affecting endocrine tissues, and manifests as the multiple endocrine tumors characteristic of Carney complex. The condition represents a significant model of how disruption in a single component of the cAMP signaling pathway can lead to system-wide pathological consequences .

Research into genotype-phenotype correlations is ongoing, with evidence suggesting that specific mutation types may correlate with particular clinical manifestations of the syndrome. Investigating these correlations requires comprehensive genetic analysis alongside detailed clinical phenotyping.

What experimental approaches are most effective for studying PKAR-I alpha protein-protein interactions?

Several complementary experimental approaches can be employed to study PKAR-I alpha protein-protein interactions:

  • Co-immunoprecipitation (Co-IP): This technique allows researchers to isolate PKAR-I alpha along with its binding partners from cell lysates using specific antibodies. The precipitated complexes can then be analyzed by western blotting or mass spectrometry to identify interacting proteins.

  • Yeast Two-Hybrid (Y2H) Assays: This system can detect direct interactions between PKAR-I alpha and potential binding partners, though it should be validated with other methods due to potential false positives.

  • Förster Resonance Energy Transfer (FRET): This technique enables researchers to study protein-protein interactions in living cells by measuring energy transfer between fluorophore-tagged proteins that are in close proximity.

  • Surface Plasmon Resonance (SPR): Using recombinant PKAR-I alpha (>95% purity), SPR can provide quantitative binding kinetics data for interactions with potential partners .

  • Protein Microarrays: These allow for high-throughput screening of multiple potential interactions simultaneously.

For optimal results, researchers should use recombinant PKAR-I alpha stored at 4°C for short-term use (2-4 weeks) or at -20°C for long-term storage, avoiding multiple freeze-thaw cycles to maintain protein integrity .

What are the optimal conditions for expressing and purifying recombinant PKAR-I alpha?

The optimal expression and purification of recombinant PKAR-I alpha typically involves:

  • Expression System: E. coli is commonly used for expressing recombinant PKAR-I alpha, as it allows for high-yield protein production. The protein can be expressed using a cDNA sequence encoding the complete PKAR-I alpha sequence .

  • Induction Conditions: Optimization of IPTG concentration, induction temperature, and duration is crucial. Lower induction temperatures (16-18°C) often yield more soluble protein.

  • Purification Strategy:

    • Initial capture using affinity chromatography (e.g., Ni-NTA for His-tagged versions)

    • Further purification using ion-exchange chromatography

    • Final polishing step with size-exclusion chromatography to achieve >95% purity

  • Buffer Composition: Optimal storage buffer contains 20mM MOPS (pH 7.0), 150mM NaCl, 1mM 2-mercaptoethanol, and 50% glycerol .

  • Quality Control: Assessment by SDS-PAGE and functional assays to confirm purity and activity.

For applications in cell culture, testing for endotoxin levels is recommended prior to use, as bacterial expression systems can introduce endotoxins that may affect cell-based assays .

How can researchers effectively design experiments to study the role of PKAR-I alpha in cAMP signaling?

An effective experimental design to study PKAR-I alpha in cAMP signaling should include:

  • Baseline Characterization: Establish baseline PKAR-I alpha expression and activity levels in your experimental system.

  • Perturbation Approaches:

    • Genetic approaches: CRISPR/Cas9-mediated knockout or knockdown of PRKAR1A

    • Pharmacological approaches: Use of cAMP analogs or phosphodiesterase inhibitors

    • Expression of mutant variants: Particularly those associated with Carney complex

  • Readout Systems:

    • Direct measurement of PKA activity using substrate phosphorylation assays

    • Visualization of signaling using FRET-based reporters

    • Downstream gene expression analysis using qRT-PCR or RNA-seq

  • Control Conditions:

    • Include wild-type PKAR-I alpha as a positive control

    • Use catalytically inactive mutants as negative controls

    • Include other PKA regulatory subunit isoforms to assess specificity

  • Temporal Considerations: Design experiments to capture both immediate and delayed responses, as cAMP signaling involves both rapid phosphorylation events and longer-term transcriptional changes.

How should researchers address data variability in PKAR-I alpha functional studies?

Researchers should implement the following approaches to address data variability in PKAR-I alpha functional studies:

  • Experimental Replication:

    • Perform at least three independent biological replicates

    • Include technical replicates within each biological experiment

    • Standardize experimental conditions across replicates

  • Statistical Analysis:

    • Apply appropriate statistical tests based on data distribution (parametric vs. non-parametric)

    • Use ANOVA with post-hoc tests for multiple comparisons

    • Report effect sizes alongside p-values

  • Normalization Strategies:

    • Normalize kinase activity to total protein concentration

    • Use internal controls such as housekeeping genes for expression studies

    • Apply batch correction methods for experiments performed on different days

  • Control Inclusion:

    • Use wild-type PKAR-I alpha as a reference point

    • Include both positive and negative controls in each experiment

    • Consider temperature, pH, and buffer composition effects on protein stability

  • Reporting Standards:

    • Document all experimental conditions thoroughly

    • Report all data points, not just averages

    • Address outliers transparently with justification for any exclusions

By implementing these approaches, researchers can enhance data reliability and facilitate cross-study comparisons in the field.

What are the key considerations when interpreting conflicting results regarding PKAR-I alpha function?

When faced with conflicting results regarding PKAR-I alpha function, researchers should consider:

  • Methodological Differences:

    • Expression systems (prokaryotic vs. eukaryotic)

    • Protein tags and their potential impact on function

    • Assay conditions and buffer compositions

    • Reaction kinetics and time points measured

  • Context-Dependent Effects:

    • Cell type-specific differences in PKAR-I alpha behavior

    • Influence of expression levels on function

    • Interaction with cell-specific binding partners

    • Post-translational modifications affecting activity

  • Technical Limitations:

    • Sensitivity and specificity of detection methods

    • Dynamic range of assays

    • Potential artifacts introduced by experimental manipulations

  • Biological Complexity:

    • Compensatory mechanisms when PKAR-I alpha is perturbed

    • Redundancy with other PKA regulatory subunits

    • Cross-talk with other signaling pathways

  • Resolution Strategies:

    • Use complementary methodological approaches

    • Collaborate with laboratories reporting different results

    • Perform side-by-side comparisons under identical conditions

    • Consider meta-analysis approaches when multiple studies exist

Understanding these factors can help researchers contextualize conflicting results and design experiments to resolve discrepancies.

How does research on PKAR-I alpha contribute to understanding the pathogenesis of Carney complex?

Research on PKAR-I alpha provides critical insights into Carney complex pathogenesis through several mechanisms:

  • Molecular Basis: Functional null mutations in PRKAR1A lead to dysregulated PKA activity, establishing a direct molecular link between this regulatory subunit and disease development .

  • Genotype-Phenotype Correlations: Different mutations in PRKAR1A may correlate with specific clinical manifestations, helping to explain the variable presentation of Carney complex.

  • Tumor Development Mechanisms: Understanding how PKAR-I alpha dysfunction leads to multiple endocrine tumors illuminates broader processes of endocrine tumorigenesis.

  • Signaling Networks: Research reveals how perturbation of a single component in the cAMP-PKA pathway can have system-wide effects, demonstrating the interconnectedness of cellular signaling networks.

  • Disease Progression: Longitudinal studies of PKAR-I alpha mutations help explain the age-dependent penetrance and variable expressivity observed in Carney complex patients.

This research has translational significance, potentially informing development of targeted therapies that could address the root cause of the disease rather than just managing individual tumors.

What are the implications of PRKAR1A gene rearrangements in thyroid carcinogenesis?

The PRKAR1A gene can fuse with the RET protooncogene through gene rearrangement, forming a thyroid tumor-specific chimeric oncogene known as PTC2 . This has several important implications for thyroid carcinogenesis:

  • Oncogenic Mechanism: The fusion creates a constitutively active signaling molecule that drives cell proliferation and tumor formation through aberrant activation of RET signaling pathways.

  • Diagnostic Value: Detection of PRKAR1A-RET fusion (PTC2) can serve as a molecular marker for specific subtypes of thyroid cancer, potentially allowing for more precise diagnosis.

  • Therapeutic Targeting: Understanding this fusion provides a potential therapeutic target, as agents that disrupt the function of the fusion protein or its downstream pathways could be developed.

  • Risk Stratification: Presence of this fusion may correlate with specific clinical outcomes, allowing for better patient stratification and treatment planning.

  • Tumorigenesis Model: This gene rearrangement provides a model for studying how disruption of normal PKAR-I alpha function contributes to cellular transformation and tumor development.

Researchers investigating this fusion should consider using modern sequencing approaches to detect the precise breakpoints and fusion architecture, which may vary between patients and affect oncogenic potential.

What novel technologies are advancing our understanding of PKAR-I alpha dynamics and interactions?

Several cutting-edge technologies are transforming PKAR-I alpha research:

  • Cryo-Electron Microscopy: Providing high-resolution structural insights into PKAR-I alpha conformational changes upon cAMP binding and interaction with catalytic subunits.

  • Single-Molecule FRET: Allowing real-time observation of PKAR-I alpha conformational dynamics in response to cAMP concentration fluctuations.

  • Optogenetic Approaches: Enabling precise temporal control of PKA activity through light-sensitive cAMP production, revealing the kinetics of PKAR-I alpha response.

  • Proximity Labeling Techniques (BioID, APEX): Identifying novel PKAR-I alpha interactors in their native cellular context.

  • Protein Engineering: Creating PKAR-I alpha variants with altered cAMP sensitivity or catalytic subunit binding properties for mechanistic studies.

  • CRISPR-Based Imaging: Visualizing endogenous PKAR-I alpha localization and dynamics in living cells.

  • Interactome Mapping: Using mass spectrometry-based approaches to comprehensively identify the PKAR-I alpha interactome under different cellular conditions.

These technologies are providing unprecedented insights into how PKAR-I alpha functions within the complex cellular signaling network and how its dysregulation contributes to disease states.

How might understanding PKAR-I alpha function contribute to therapeutic development for PKA-related disorders?

Understanding PKAR-I alpha function could inform therapeutic development through several approaches:

  • Small Molecule Modulators: Development of compounds that selectively modulate PKAR-I alpha binding to cAMP or catalytic subunits, potentially restoring normal PKA signaling in disease states.

  • Gene Therapy Approaches: For Carney complex caused by PRKAR1A mutations, gene replacement or editing strategies could restore normal regulatory function.

  • Peptide-Based Inhibitors: Design of peptides that mimic binding interfaces between PKAR-I alpha and its interaction partners to selectively disrupt pathological interactions.

  • Allosteric Modulators: Identification of compounds that bind to allosteric sites on PKAR-I alpha to fine-tune its regulatory function.

  • Targeted Protein Degradation: Development of proteolysis-targeting chimeras (PROTACs) to selectively degrade mutant PKAR-I alpha proteins while sparing wild-type function.

  • Combination Therapies: Strategies that target both PKAR-I alpha and downstream effectors to comprehensively address dysregulated signaling.

Product Science Overview

Introduction

Protein Kinase A (PKA) is a crucial enzyme in cellular signaling pathways, playing a significant role in regulating various physiological processes. The regulatory subunit-1 alpha (RIα) of PKA, encoded by the PRKAR1A gene, is essential for the proper functioning of this enzyme. This article delves into the background, structure, function, and clinical significance of the human recombinant form of PKA regulatory subunit-1 alpha.

Structure and Function

PKA is a holoenzyme composed of two regulatory subunits and two catalytic subunits. The regulatory subunits control the activity of the catalytic subunits by binding to them and keeping them inactive. The RIα subunit, specifically, is one of the regulatory subunits that play a pivotal role in this regulation .

The PRKAR1A gene provides instructions for making the RIα subunit. When cyclic AMP (cAMP) levels rise in the cell, cAMP binds to the RIα subunits, causing a conformational change that releases the catalytic subunits. These free catalytic subunits then phosphorylate various target proteins, leading to a cascade of cellular responses .

Clinical Significance

Mutations in the PRKAR1A gene can lead to several disorders. One of the most notable conditions associated with PRKAR1A mutations is Carney Complex (CNC), a multiple neoplasia syndrome characterized by abnormal skin pigmentation, myxomas, and endocrine tumors . Additionally, inactivating mutations in PRKAR1A can cause primary pigmented nodular adrenocortical disease (PPNAD), which is associated with Cushing’s syndrome .

Research and Applications

Research has shown that the depletion of RIα in mammalian cells activates the mechanistic target of rapamycin (mTOR) pathway, leading to autophagic deficiency . This finding has significant implications for understanding the molecular basis of diseases associated with autophagic deficiency, such as certain cancers and neurodegenerative disorders.

The human recombinant form of RIα is used in various research applications to study its role in cellular signaling and disease mechanisms. By using recombinant proteins, researchers can investigate the specific functions and interactions of RIα in a controlled environment, leading to a better understanding of its role in health and disease.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.